In this age of promise of new therapies for cancer, immunotherapy is emerging as an exciting treatment option for patients. Vaccines and cytokines are being tested extensively in clinical trials, and strategies using monoclonal antibodies and cell transfer are mediating dramatic regression of tumors in patients with certain malignancies. However, although initially advocated as being more specific for cancer and having fewer side effects than conventional therapies, it is becoming increasingly clear that many immunotherapies can lead to immune reactions against normal tissues. Immunotoxicities resulting from treatment can range from relatively minor conditions, such as skin depigmentation, to severe toxicities against crucial organ systems, such as liver, bowel, and lung. Treatment-related toxicity has correlated with better responses in some cases, and it is probable that serious adverse events from immune-mediated reactions will increase in frequency and severity as immunotherapeutic approaches become more effective. This review introduces immunotherapeutic approaches to cancer treatment, provides details of toxicities arising from therapy, and discusses future potential ways to avoid or circumvent these side effects.

Immunotherapy holds much promise for the treatment of cancer. The immune system is capable of dramatic and decisive responses against infectious disease, which is accomplished with exquisite specificity against antigen. Components of immunity are seen as potentially more specific weapons to direct against tumors than chemotherapy or radiation. With our expanding knowledge of tumor-associated antigens (TAAs), there are many different approaches being developed to direct immunity against transformed cells.

Immunotherapies may involve the active generation of immunity to TAA, via vaccination with peptides or peptide-pulsed dendritic cells.1  In addition, administration of immune modulators, such as cytokines, can boost existing antitumor immunity and target immune effector cells to sites of tumor growth.2  Monoclonal antibodies harness both innate and adaptive immune mechanisms and direct them against tumor cells.3  In addition, the effector functions of cytotoxic T lymphocytes have proven them to be particularly useful in targeting TAA in adoptive immunotherapeutic protocols.4 

Some TAAs are tumor specific, whose expression is entirely limited to tumors, examples of which include viral antigens expressed on cells in which viral oncogenes have contributed to cellular transformation. In these cases, immunotherapy can be used with fine specificity and very little toxicity against normal tissues.5  However, most TAAs are expressed by some cells of normal tissues and the potential exists for on-target toxicity against these tissues. These on-target toxicities can be assigned to 2 broad categories. First, they can comprise “true” autoimmunity, involving a fundamental induction of endogenous immunity against self-antigens, and we refer to this type as “autoimmunity.” Second, they can be more “drug-like” in nature, where damage is mediated directly by the immunomodulatory agent, and these toxicities are referred to as “immune-mediated.” Toxicities have been described in a proportion of patients using a range of immunotherapeutic approaches. As immune-based cancer therapies become more potent, it is probable that autoimmune and immune-mediated toxicities will become more severe. Indeed, these toxicities can be associated with better antitumor responses resulting from immunotherapy.

In this review, we introduce various immunotherapeutic approaches and provide details of toxicity to normal tissues resulting from these approaches, as well as describe potential ways to overcome these toxicities.

Adoptive immunotherapy is a very promising approach to treating cancer that involves the isolation of leukocytes and their activation and expansion in vitro followed by infusion into patients. Advantages of this type of approach include the opportunity to manipulate and activate lymphocytes away from the in vivo immunosuppressive environment, and their expansion to vast numbers, thereby circumventing many regulatory checkpoints and delivering “instant” immunity. Adoptive immunotherapy has been demonstrated to induce regression of established tumors, often complete, in both mouse models of disease and in patients.4  However, apart from Epstein-Barr virus–associated malignancies, this form of therapy targets antigens expressed on some normal tissues besides tumor cells, and immune-mediated toxicity has been observed in the treatment of both mice and humans (Table 1; Figure 1).

Table 1

Immune-mediated toxicities associated with adoptive immunotherapy of cancer

Cell typeSpeciesTarget antigenTumorToxicitiesReference(s)
CTL Mouse (or rat) Pnma-1 Paraneoplastic syndrome (rat) CNS inflammation 80  
CTL Recoverin Fibrosarcoma Retinal dysfunction 81  
CTL with IL-2 and vaccine Various Melanoma Melanocyte destruction, ocular toxicity 8,82  
CD4+ T cells TRP-1 Melanoma Vitiligo 83,84  
Immune T cells CEA Colon Colitis 85  
Transgenic T cells Telomerase Prostate Reduction in B cells 86  
TCR transgenic T cells Various None Pancreatitis, colitis 11  
CAR-modified T cells CD19 Lymphoma Depletion of normal B cells 87,88  
CAR-modified T cells VEGF-R2 Various Various organs, probably the result of cytokine-induced hypotension 89  
TIL and IL-2 Human Various Melanoma Autoimmune thyroiditis, systemic and ocular autoimmunity 90,92  
TIL with lymphodepletion and IL-2 Various Melanoma Vitiligo, uveitis 9,93  
CTL MART-1 Melanoma Melanocyte destruction 94  
TCR gene–modified T cells MART-1, gp100 Melanoma Melanocyte destruction 10  
CAR-modified T cells CAIX RCC Liver toxicity (grade 2-4) in 3 of 7 patients; lower grade in 4 of 7 patients with reduced dosing 14  
CAR-modified T cells CD19 Lymphoma B-cell depletion 15  
CAR-modified T cells Her-2 Colorectal cancer Lung toxicity 17  
CAR-modified T cells CEA Colorectal cancer Colitis 19  
Cell typeSpeciesTarget antigenTumorToxicitiesReference(s)
CTL Mouse (or rat) Pnma-1 Paraneoplastic syndrome (rat) CNS inflammation 80  
CTL Recoverin Fibrosarcoma Retinal dysfunction 81  
CTL with IL-2 and vaccine Various Melanoma Melanocyte destruction, ocular toxicity 8,82  
CD4+ T cells TRP-1 Melanoma Vitiligo 83,84  
Immune T cells CEA Colon Colitis 85  
Transgenic T cells Telomerase Prostate Reduction in B cells 86  
TCR transgenic T cells Various None Pancreatitis, colitis 11  
CAR-modified T cells CD19 Lymphoma Depletion of normal B cells 87,88  
CAR-modified T cells VEGF-R2 Various Various organs, probably the result of cytokine-induced hypotension 89  
TIL and IL-2 Human Various Melanoma Autoimmune thyroiditis, systemic and ocular autoimmunity 90,92  
TIL with lymphodepletion and IL-2 Various Melanoma Vitiligo, uveitis 9,93  
CTL MART-1 Melanoma Melanocyte destruction 94  
TCR gene–modified T cells MART-1, gp100 Melanoma Melanocyte destruction 10  
CAR-modified T cells CAIX RCC Liver toxicity (grade 2-4) in 3 of 7 patients; lower grade in 4 of 7 patients with reduced dosing 14  
CAR-modified T cells CD19 Lymphoma B-cell depletion 15  
CAR-modified T cells Her-2 Colorectal cancer Lung toxicity 17  
CAR-modified T cells CEA Colorectal cancer Colitis 19  

CTL indicates cytotoxic T lymphocyte; CNS, central nervous system; IL-2, interleukin-2; and TIL, tumor-infiltrating lymphocytes.

Figure 1

Immune toxicity associated with immunotherapy of cancer. Therapeutic strategies, including vaccines, adoptive immunotherapy, cytokines, and antibodies, can induce immunity against tumor antigens. However, these immune responses can also cause damage to a variety of organ systems as shown.

Figure 1

Immune toxicity associated with immunotherapy of cancer. Therapeutic strategies, including vaccines, adoptive immunotherapy, cytokines, and antibodies, can induce immunity against tumor antigens. However, these immune responses can also cause damage to a variety of organ systems as shown.

Close modal

Tumor-infiltrating lymphocytes and melanoma

Tumor inhibition has been described using adoptive immunotherapy in various animal models over the past 50 years, but antigen specificity and its expression on normal tissue were largely undefined in earlier models.6,7  More recently, mouse tumor models with known tumor antigens also expressed on self-tissues have become available and effective antitumor responses after adoptive immunotherapy has been observed.8  However, immune-mediated toxicity has been observed in these mouse models targeting normal tissues, including skin, eye, colon, and the B-cell compartment as summarized in Table 1. In the following discussion, we focus on these toxicities following adoptive immunotherapy in humans.

One of the most promising applications of adoptive immunotherapy in the clinic involves the use of tumor-infiltrating lymphocytes to treat melanoma. Lymphocytes derived from tumors can be expanded to yield many billions of cells that are reactive with a range of melanoma antigens, including gp100, MART-1, and tyrosinase.

After demonstrations that prior lymphodepletion could lead to enhanced persistence and activity of transferred T cells, adoptive immunotherapy protocols were modified to include preconditioning regimens to deplete cells of the hematopoietic system. Depletion regimens ranged from nonmyeloablative, using cyclophosphamide and fludarabine, to fully ablative regimens using chemotherapy and whole body irradiation, together with stem cell support. Objective response rates of adoptive immunotherapy reached 70% with these modifications, but immune-mediated toxicities were also observed, with reports of vitiligo and occasional reports of ocular toxicity, involving responses against melanin-containing cells important in retina function.9 

In cases where endogenous tumor-infiltrating lymphocytes are difficult to obtain, tumor-specific T cells can be generated from peripheral blood lymphocytes by genetic modification with genes encoding specific T-cell receptor (TCR) α- and β-chains. In a 2009 trial, melanoma patients treated with gene-modified T cells reactive with melanoma/melanocyte antigens experienced destruction of normal melanocytes in the skin (27 of 36 patients), as well as responses against normal cells of the eye and ear, which occurred in approximately 50% of 20 patients receiving T cells modified with a highly reactive TCR.10  These patients were administered steroids to inhibit T-cell activity, which led to resolution of both uveitis and hearing loss.

Interestingly, in addition to the aforementioned on-target immune toxicities, the potential for immune-mediated off-target toxicity using adoptive transfer of TCR gene-modified T cells has been demonstrated in mice. In a study using mouse T cells genetically engineered to express TCR α- and β-chains specific for ovalbumin, severe toxicity was observed, including cachexia, anemia, pancreatitis, and colitis.11  Reactivity against these normal tissues was thought to be the result of mispairing of introduced and endogenous TCR chains leading to T cells with neo-specificities. These findings were also extended to include T cells modified to express other TCRs, including those specific for gp100, simian virus 40 large T antigen, TRP-2, and influenza virus nucleoprotein, although the incidence of lethal toxicity varied depending on the TCR. The propensity of TCR mispairing to induce immune-mediated toxicity in humans has yet to be determined fully, but it has not been observed to date in clinical trial.12 

Genetically redirected T cells in adoptive immunotherapy

By far the greatest application of adoptive immunotherapy has been in the melanoma setting as described in “Tumor-infiltrating lymphocytes and melanoma.” This is largely because of the availability of specific T cells, and extension to other common cancers is restricted by a lack of availability of endogenous T cells of appropriate specificity. However, T cells reactive with a range of common cancers can be generated by genetic modification of peripheral blood lymphocytes with chimeric antigen receptors (CARs), whose specificity is derived from monoclonal antibodies specific for cell surface TAA. CAR-modified T cells have been used in clinical trials for a range of cancers, including ovarian cancer, neuroblastoma, colon cancer, and lymphoma.13  The use of CAR-modified T cells is in its infancy, and only limited antitumor effects have been described to date. Nevertheless, in a phase 1 study for renal cell carcinoma (RCC) targeting the TAA carbonic anhydrase IX, adoptive transfer of gene-modified T cells led to grade 3 to 4 liver toxicity in 3 of 7 patients treated. Toxicity was thought to be the result of T cells targeting the CAIX antigen also present on bile ducts.14  Toxicity was resolved in this study after cessation of adoptive T-cell transfer or administration of steroids.

CAR-modified T-cell activity against normal cells was also observed in a clinical study targeting CD19 for the treatment of follicular lymphoma.15  The CAR was composed of a single-chain anti-CD19 antibody linked to CD28 and the ζ-chain of the CD3-TCR complex. In this study, dramatic regression of malignant cells was observed, but a prolonged depletion of normal B cells was also observed, leading to greatly decreased levels of serum immunoglobulin. Although low levels of serum antibody are concerning, administration of exogenous immunoglobulin can correct for this deficiency, thereby providing protection against infection. In another study targeting CD19 with CAR-modified T cells, this time for chronic lymphocytic leukemia, treatment was well tolerated in 3 patients receiving T-cell transfer in the absence of prior lymphodepletion, with only transient fevers experienced. However, a patient receiving T cells after lymphodepletion developed hypotension, dyspnea, and renal failure and died 4 days after treatment.16  Death in this case was not thought to be the result of treatment and was attributed to sepsis because of infection.

More recently, adoptive transfer of CAR-modified T cells, specific for the TAA human epidermal growth factor receptor-2 (Her-2), led to death of a patient receiving treatment for colon cancer.17  Toxicity involved respiratory distress and may have been the result of CAR-expressing T-cell activity against normal Her-2–expressing cells of the lung. Increased levels of serum cytokines, including IFN-γ, GM-CSF IL-6, and TNF-α, were observed, together with hypotension, brachycardia, and gastrointestinal bleeding, which led to cardiac arrest. In another study targeting Her-2, no toxicity was observed,18  although comparisons between this study using autologous cytotoxic T lymphocyte clones and the CAR-expressing study are difficult because of significant differences in the studies, including the use of lymphodepleting conditioning and greater numbers of T cells in the CAR study. Thus, many different organ systems can become targets of immune-mediated damage after adoptive immunotherapy, a recent example of which involved severe, yet transient, colitis in 3 of 3 patients receiving T cells specific for the colon cancer-associated antigen, carcinoembryonic antigen.19 

In the aforementioned examples, the target tumor antigens were also expressed by a range of normal tissues. However, antigens with greater tumor specificity are emerging as more desirable targets. The cancer/testes antigen, NY-ESO-1, represents such a target that is expressed on a range of tumors, but whose normal tissue expression is limited to testes.20  In a phase 1 study, > 50% of patients with either melanoma or synovial cell sarcoma experienced objective clinical responses in the absence of any immune-mediated toxicity.21  Similarly, in a phase 1 study targeting the carbohydrate antigen GD2 with genetically redirected T cells, no T cell–related adverse events were observed, despite apparent antitumor activity demonstrated in 4 of 8 evaluable patients.22  GD2 is an attractive target because it is expressed on neuroblastoma and many melanomas, but normal tissue expression is largely limited to brain and peripheral nerves.

There is much promise and excitement in the use of monoclonal antibodies for immunotherapy, with 10 approved by the Food and Drug Administration for the treatment of cancer (reviewed by Weiner et al23 ). These antibodies are specific for a variety of molecular targets expressed on a range of cancers, including lymphomas, leukemias, breast cancer, and colorectal cancer. A variety of effector mechanisms are used by antibodies against tumor cells, which include antagonizing growth factors and their receptors, or inducing their degradation. Alternatively, antibodies may activate antibody-dependent cell-mediated cytotoxicity (ADCC) or the complement pathway. Finally, antibodies may also be used to antagonize receptors, such as cytotoxic T lymphocyte antigen-4 (CTLA-4), which normally down-regulate immune responses.

Clinical trials are currently under way to test the potential of antibodies to mediate tumor regression. However, just as spontaneously arising tumor-specific antibodies have been shown to induce autoimmune pathologies in paraneoplastic neurologic disorders,24  toxicities against normal tissues have also been observed in a proportion of patients receiving exogenous antibody (Table 2).

Table 2

Immune-mediated toxicities associated with antibody therapy of cancer

Antibody regimenTarget antigenCancer typeToxicitiesReference(s)
Ipilimumab CTLA-4 Hodgkin disease, myeloma, AML, CML, CLL, NHL Arthritis, hyperthyroidism, pneumonitis 29  
Ipilimumab CTLA-4 Melanoma Colitis, bowel perforation, vitiligo, hypophysitis 33  
Tremelimumab or ipilimumab CTLA-4 Melanoma Colitis, dermatitis 30,32,95  
Anti-CTLA-4 + TAA peptides CTLA-4 Melanoma Colic, dermatitis, uveitis, enterocolitis, hepatitis, hypophysitis, vitiligo, pulmonary leukocyte infiltration 28,96,,99  
Anti-CTLA-4 (with vaccines and cytokines) CTLA-4 Melanoma (mouse and human) Melanocyte destruction, enteritis 31,100,102  
Anti-CTLA-4, irradiated tumor cells, GM-CSF CTLA-4 C2 prostate cancer (mouse) Prostatitis, destruction of prostate epithelium 103  
MDX-1106 PD-1 Melanoma, RCC, NSCLC, prostate Colitis 34  
Alemtuzumab CD52 CLL Destruction of normal leukocytes, leading to susceptibility to infections 42,44,104  
Gemtuzumab CD33 AML Infection, sepsis, pneumonia 36,38  
Rituxumab CD20 Lymphoma Suppression of B cells leading to deficiency in immunoglobulin and infections 48,49,105,106  
Antibody regimenTarget antigenCancer typeToxicitiesReference(s)
Ipilimumab CTLA-4 Hodgkin disease, myeloma, AML, CML, CLL, NHL Arthritis, hyperthyroidism, pneumonitis 29  
Ipilimumab CTLA-4 Melanoma Colitis, bowel perforation, vitiligo, hypophysitis 33  
Tremelimumab or ipilimumab CTLA-4 Melanoma Colitis, dermatitis 30,32,95  
Anti-CTLA-4 + TAA peptides CTLA-4 Melanoma Colic, dermatitis, uveitis, enterocolitis, hepatitis, hypophysitis, vitiligo, pulmonary leukocyte infiltration 28,96,,99  
Anti-CTLA-4 (with vaccines and cytokines) CTLA-4 Melanoma (mouse and human) Melanocyte destruction, enteritis 31,100,102  
Anti-CTLA-4, irradiated tumor cells, GM-CSF CTLA-4 C2 prostate cancer (mouse) Prostatitis, destruction of prostate epithelium 103  
MDX-1106 PD-1 Melanoma, RCC, NSCLC, prostate Colitis 34  
Alemtuzumab CD52 CLL Destruction of normal leukocytes, leading to susceptibility to infections 42,44,104  
Gemtuzumab CD33 AML Infection, sepsis, pneumonia 36,38  
Rituxumab CD20 Lymphoma Suppression of B cells leading to deficiency in immunoglobulin and infections 48,49,105,106  

CTLA-4 indicates cytotoxic T lymphocyte antigen-4; AML, acute myeloid leukemia; CML, chronic myeloid leukemia; CLL, chronic lymphocytic leukemia; NHL, non-Hodgkin lymphoma; and NSCLC, non–small cell lung cancer.

Toxicities arising from antibody administration can occur in various ways. First, toxicity can follow the induction of potent endogenous autoimmunity against both tumor antigens and other self-antigens, resulting in both on- and off-target toxicities. Second, toxicities can involve on-target depletion of normal cell subsets, compromising normal tissue function. In this case, cell depletion and resulting toxicity are generally transient, persisting only during antibody administration, after which normal cells are replaced from precursors. Nevertheless, even transient depletion can have severe consequences.

Some therapeutic antibodies mediate their antitumor activity by blocking growth factor receptors. Antibodies in this category include bevacizumab, cetuximab, and trastuzumab, which target VEGF-R, EGF-R, and Her-2, respectively. Toxicities against a range of normal tissues, including gastrointestinal tract, skin, and lung, have been observed using these antibodies for cancer therapy. However, because these antibodies mediate their effects largely by blocking the signaling ability of growth factor receptors, which is reminiscent of the action of drugs rather than immunity, we have not discussed these antibodies here but cite references as information for the reader.25-27 

Autoimmunity involving on- and off-target toxicities

An immune response is a complex process that is subject to many controls and balances, including the involvement of inhibitory molecules on lymphocytes that down-regulate responses (on elimination of antigen) or prevent inappropriate activity against self-antigens. CTLA-4 is one such regulatory molecule expressed by T cells that transmits an inhibitory signal to T cells on binding to CD80 and CD86 on antigen-presenting cells. The targeting of this inhibitory receptor in immunotherapy has been used to break immune tolerance of T cells to TAAs, resulting in the expansion of T cells that elicit antitumor effects.28  However, in addition to tumor regression, anti–CTLA-4 antibodies, such as ipilimumab and tremelimumab, have been associated with autoimmunity affecting tissues, including the thyroid, lung, joints, gastric mucosa, and liver (Table 2). In a clinical study using ipilimumab to treat hematopoietic malignancy after recurrence after allogeneic hematopoietic cell transplantation, complete remissions were observed in 2 Hodgkin disease patients (2 of 29) and a partial remission in a patient with mantle cell lymphoma. However, grade 2 to 4 autoimmune events, thought to be treatment-related observed in 4 patients in this study, included arthritis, hyperthyroidism, and recurring pneumonitis.29 

Toxicities can be common and sometimes severe, as seen in a study using ipilimumab alone or together with vaccine for the treatment of melanoma or RCC. In this study, 21% of 198 patients developed grade 3 to 4 enterocolitis, with 4 patients having colonic perforation leading to 2 deaths and one colectomy.30  Enterocolitis resolved in most patients after administration of corticosteroids, providing further evidence of immune involvement. However, it was not clear whether lymphocyte responses were directed against endogenous antigens from normal bowel or antigens derived from intestinal microflora. Gastrointestinal toxicity was also observed in another study, where 11 of 71 patients receiving ipilimumab developed grade 3 to 4 diarrhea, and gastrointestinal bleeding required colectomy in one patient.31 

Of significant interest is that autoimmunity has been demonstrated to be associated with clinical response, suggesting that the greater the immune dysregulation mediated by anti–CTLA-4, the greater the antitumor effect. In some of these cases, specific T cells have been identified that were associated with both antitumor activity and autoimmunity, where Melan-A–specific cytotoxic T cells were observed infiltrating tumors and skin rashes together with a 30-fold increase in circulating Melan-A–specific cytotoxic T cells after treatment with ipilimumab.32 

Further randomized clinical studies continue to determine whether the benefits of anti–CTLA-4 therapy justify the risk of autoimmune side effects. In a recent phase 3 study involving 676 melanoma patients, ipilimumab (with or without gp100 vaccine) was demonstrated to significantly improve survival of patients compared with patients receiving vaccine alone (10.1 months median survival vs 6.4 months).33  Complete or partial tumor responses were observed in 9.4% of patients receiving treatment that included ipilimumab compared with 1.5% responses in patients not receiving the antibody. Immune-related adverse events occurred in 60% of ipilimumab-treated patients, with approximately 15% of patients experiencing grade 3 or 4 immune-related adverse events. Toxicity was largely reversed with appropriate treatment, although 7 deaths occurred that were thought to be immune-related because of conditions, including colitis, bowel perforation, liver failure, and 1 patient with Guillain-Barré syndrome.

A range of measures can be taken to manage immune-mediated toxicities, including the use of corticosteroids, which can decrease the severity of toxicity. Interestingly, antitumor responses have been observed to be maintained in the presence of steroid treatment, suggesting that antitumor activity can be separated from autoreactivity, at least in some circumstances.30,31 

Anti–CTLA-4 antibodies have induced antitumor responses most markedly against RCC and melanoma, which suggests that these 2 cancers are exceptionally immunogenic.28  Induction of autoimmunity by anti–CTLA-4 may occur through dysregulation of a preexisting immune response to self-antigens, which was held in check by CTLA-4 or through a de novo expansion of self-reactive T cells from naive precursors in the absence of the CTLA-4 checkpoint.

Immune checkpoints are attractive targets for immunotherapies, and much interest is being expressed in targeting immune inhibitory pathways other than CTLA-4. Programmed death-1 (PD-1) is expressed by activated T cells, and its ligand is expressed by a range of cell types, including antigen-presenting cells and sometimes by tumor cells themselves. Interaction of PD-1 with its ligand inhibits T-cell responses and down-regulates immunity.

In a clinical study, MDX-1106, a PD-1–blocking antibody, was used for the treatment of a range of malignancies, including melanoma, renal cell carcinoma, non–small cell lung carcinoma, and prostate cancer. Results from the first 39 patients indicate that 3 patients had objective tumor responses.34  Treatment was well tolerated, with a single case of colitis the only grade 3 to 4 adverse event reported potentially related to antibody administration. A more detailed knowledge of immune checkpoints may lead to effective and safe immunotherapies.

On-target toxicities from depletion of normal cell subsets

To apply effector mechanisms of ADCC and complement against malignant cells, the identification of cell surface markers is necessary. However, most molecular targets of antibodies against hematologic cancers are lineage-specific rather than tumor-specific; consequently, immune-mediated toxicity is induced against normal blood cells, which can compromise immunity or hematopoiesis. Fortunately, target markers can sometimes be selected that are not expressed on stem cells or lineage precursors; therefore, normal cells can often be reconstituted after cessation of therapy. Nevertheless, toxicities can occur because of lowered immunity after immune-mediated depletion of normal leukocytes, as seen when using antibodies for the treatment of some hematologic malignancies.

Gemtuzumab ozogamicin (Mylotarg) is an antibody-calicheamicin conjugate that targets the CD33 surface antigen. Gemtuzumab treatment has been successful in the treatment of CD33-positive AML, after internalization of the conjugate.35  However, the expression of CD33 on normal cells (myeloid progenitor cells and monocytes) has resulted in a large proportion of patients experiencing neutropenia (> 90%) resulting in pneumonia and infections (∼ 20%), which can be life-threatening.36,37  Another serious effect of gemtuzumab is the development of hepatotoxicity, such as vascular obstructive disease, which damages the hepatic sinusoidal epithelium.38  This is thought to occur via receptor-mediated uptake of the antibody-calicheamicin complex through CD33 expression on liver cell populations, such as Kupffer cells (up to 48% of patients in some studies).39  Although CD33 is still a target of interest for treating acute myeloid leukemia and some patients can benefit from treatment with gemtuzumab,40  Mylotarg was voluntarily withdrawn from the market in June 2010 because no significant benefit of the antibody conjugate in combination with chemotherapy was demonstrated above chemotherapy alone.

Alemtuzumab (MabCampath) provides another example where subsets of normal leukocytes are depleted after treatment of hematologic malignancy. Alemtuzumab is used in the treatment of CLL and binds to CD52, which is present on both normal and malignant B and T lymphocytes, as well as monocytes, macrophages, and eosinophils.41  The use of alemtuzumab has yielded promising results, with objective response rates of up to 90% in CLL and encouraging results against other malignancies, such as T-cell prolymphocytic leukemia and peripheral T-cell lymphoma.42,43  As a result of expression of CD52 on normal leukocytes, alemtuzumab also depletes normal immune cell populations, which can result in severe infections that can be fatal.42,44 

On-target depletion of normal leukocytes has also been observed using rituximab, which is specific for CD20 expressed on a variety of non-Hodgkin lymphomas and normal mature B cells.45  Rituximab has been used in the treatment of malignancies, including follicular and diffuse large B-cell lymphoma.46,47  It has been shown to mediate lysis of lymphoma cells through a complement-dependent mechanism, and it has substantially increased the survival rates of patients with B-cell lymphomas since its approval by the FDA in 1997. However, the use of this antibody has been associated with toxicities, including tumor lysis syndrome, infections, and reactivation of viruses, such as hepatitis B and herpes simplex virus.48,49  Although antibody-producing plasma cells are not depleted with rituximab, several other cell subsets, including mature B cells, are depleted, which is thought to be responsible for increased rates of infection. Interestingly, depletion of neutrophils has also been observed after the use of rituximab.50  The extent of neutropenia did not generally have clinical significance in these cases; and although the mechanism of neutrophil depletion was not clear, the production of antineutrophil antibodies and disruption of neutrophil homeostasis resulting from B-cell depletion have been proposed as potential contributing causes.

A range of cytokines are being tested in the clinic for their ability to recruit immunity against tumor cells by inducing the activation, proliferation, and survival of tumor-specific lymphocyte populations.2  The complexity of cytokine interactions with the immune system means that often the outcomes of their use cannot be fully predicted. It is therefore not entirely unexpected that autoimmune pathologies have been encountered in a number of cytokine trials to date (Table 3).

Table 3

Autoimmunity associated with cytokine administration in cancer in humans

Cytokine administeredCancer targetedAutoimmune eventsReference(s)
IFN-α Chronic myeloid leukemia Thyroditis, sarcoidosis, systemic lupus erythematosus associated with autoantibodies, autoimmune thrombocytopenic purpura, acute pancreatitis, hyperthyroidism, aggravation of psoriasis 51,107,,110  
IFN-α Cutaneous T-cell lymphoma Psoriasis 111  
IFN-α Mid-gut carcinoid tumors Systemic lupus erythematosus, pernicious anemia, thyroid disease 112  
IFN-α2b Melanoma Hypothyroidism 113  
IFN-α2b and piroxicam Melanoma Vitiligo, pulmonary interstitial fibrosis 114  
GM-CSF with gp100 and tyrosinase peptides Melanoma Vitiligo 115  
IL-2 RCC Hypothyroidism, myasthenia gravis, diabetes mellitus (insulin-dependent), and myositis 62,64,116  
IL-2 Melanoma Vitiligo 67  
IL-2 Colorectal cancer Diabetes mellitus 117  
IL-2 and IFN-γ RCC Autoantibodies against erythrocytes 65  
IL-2 and IFN-α2b RCC, melanoma Antithyroid antibodies, hypothyroidism, autoimmune thyroiditis 63,118,119  
Cytokine administeredCancer targetedAutoimmune eventsReference(s)
IFN-α Chronic myeloid leukemia Thyroditis, sarcoidosis, systemic lupus erythematosus associated with autoantibodies, autoimmune thrombocytopenic purpura, acute pancreatitis, hyperthyroidism, aggravation of psoriasis 51,107,,110  
IFN-α Cutaneous T-cell lymphoma Psoriasis 111  
IFN-α Mid-gut carcinoid tumors Systemic lupus erythematosus, pernicious anemia, thyroid disease 112  
IFN-α2b Melanoma Hypothyroidism 113  
IFN-α2b and piroxicam Melanoma Vitiligo, pulmonary interstitial fibrosis 114  
GM-CSF with gp100 and tyrosinase peptides Melanoma Vitiligo 115  
IL-2 RCC Hypothyroidism, myasthenia gravis, diabetes mellitus (insulin-dependent), and myositis 62,64,116  
IL-2 Melanoma Vitiligo 67  
IL-2 Colorectal cancer Diabetes mellitus 117  
IL-2 and IFN-γ RCC Autoantibodies against erythrocytes 65  
IL-2 and IFN-α2b RCC, melanoma Antithyroid antibodies, hypothyroidism, autoimmune thyroiditis 63,118,119  

GM-CSF indicates granulocyte-macrophage colony-stimulating factor; and IL-2, interleukin-2.

IFN-α

IFN-α is a prominent and sometimes first-line therapy against many cancers, including melanoma, RCC, cutaneous T-cell lymphoma, chronic myeloid leukemia, bladder cancer, and ovarian cancer. The receptor for IFN-α is almost ubiquitously expressed and can signal to leukocytes and tumors in a variety of ways. It can have pro- or antiproliferative effects, pro- or antiapoptotic effects, modulate cell immunogenicity, promote immune responses, and inhibit angiogenesis.

An extensive range of autoimmune complications have been reported after IFN-α therapy for cancer (Table 3).51  Adverse events include diabetes and vitiligo, as well as the aggravation of preexisting autoimmune diseases. Prospective studies have indicated that autoimmunity in patients correlates with prolonged relapse-free and overall survival.52  This provides evidence that in humans IFN-α is working against cancer, at least in part, by boosting the immune response. The mechanisms by which IFN-α induces cancer regression remain the subject of ongoing investigation. It has been shown to act both on host leukocytes and directly on tumor cells.53,54 

Both B and T cell–mediated autoimmunities have been reported in response to IFN-α, which is consistent with known activities of type I interferons. Type I interferons can promote skewing of T cells to a cytotoxic phenotype by stimulating their expression of the IL-12R.55  They complement this by also inducing genes coding for MHC-I and TAAs on tumor and nontumor populations.56  IFN-α may indirectly promote the proliferation of memory T-cell populations57  and also promotes B-cell switching to the IgG2a isotype. IgG2a is capable of targeting cells for phagocytosis and triggering complement-mediated cytotoxicity.58 

In summary, type I IFNs are powerful modulators of antitumor immunity. The information available regarding their effects on leukocytes, host stroma, and tumor cells serves as a further example of the pleiotropic effects that cytokines can mediate.

IL-2

IL-2 has been widely applied in the clinic in the treatment of melanoma, RCC, and AML and is one of only a few cytokines that has progressed to FDA approval in the treatment of cancer. Used alone in the treatment of metastatic melanoma, it has produced objective response rates of up to 16%.59  The antitumor effect of IL-2 stems, at least in part, from its ability to act as a growth factor for T cells and to enhance the cytolytic activity of NK cells.

Nonimmune systemic toxicities associated with high-dose IL-2 therapy are well appreciated. IL-2 therapy induces systemic inflammation, the exact mechanism of which is poorly understood, and the main symptomatic outcomes are hypotension and capillary leak syndrome, accompanied by flu-like symptoms, vomiting, and diarrhea. Despite its toxicity, advances in managing symptoms and the resolution of toxicity after cessation of administration have led to its continued use in the cancer setting.60,61 

In addition to these systemic toxicities, IL-2 therapy is known to both exacerbate autoimmunity and trigger it de novo (Table 3). In a notable case study, both of these outcomes were detected within the same patient after high-dose IL-2 therapy for RCC, with worsening of diabetes and induction of myasthenia gravis and polymyositis.62  There is evidence that IL-2 can augment humoral responses specific for autoantigens and drive expansion of NK and T cells.63-65  Thus, both humoral and cell-mediated immune mechanisms may contribute to autoimmunity resulting from cytokine therapy.66  Interestingly, similar to observations using antibodies or adoptive immunotherapy for cancer treatment as discussed in “Autoimmunity associated with antibody therapy,” antitumor responses in patients receiving cytokines can correlate with autoimmunity.63,64 

The best evidence that high-dose IL-2 can induce on-target (tumor antigen) autoimmunity stems from a prospective study of patients treated for metastatic melanoma and RCC.67  Here, vitiligo (autoimmune destruction of melanocytes) was observed in 11 of 74 of patients treated for melanoma but 0 of 104 treated for RCC. Development of vitiligo in melanoma patients correlated with objective responses to therapy, indicating that immune targeting of melanoma differentiation antigens was underpinning both outcomes. Other autoimmune toxicities reported in conjunction with IL-2 therapy include diabetes mellitus and hypothyroidism (Table 3). These are presumed to represent examples of off-target autoimmunity in the absence of reported antigen sharing between tumors and the affected tissues.

Vaccines are a major focus of cancer immunotherapy, and lessons learned from successful vaccines against infectious disease are being applied to the treatment of malignant disease. Antigen formulations used in cancer vaccines include peptides and whole proteins in combination with adjuvants to improve immunogenicity or pulsed directly onto dendritic cells to facilitate antigen presentation. In addition, antigen has been encoded in recombinant viruses in attempts to generate robust immune responses to TAAs in parallel to viral antigens.

The vaccination approach has shown considerable promise in preclinical murine models, although breaking of tolerance toward TAAs expressed by both the tumor and healthy peripheral tissues of mice has been observed to result in autoimmune disease in some cases (summarized in Table 4). In humans, cancer vaccines have been studied intensively, particularly in the melanoma setting. Increased frequencies of antigen-specific circulating T cells have often been observed after vaccine administration, although overall clinical responses rates of only 2.6% have been achieved.68  Perhaps in keeping with the low response rate, there have been few reports of autoimmunity arising in patients after vaccination with TAAs. An evaluation of autoimmunity in melanoma patients treated with IL-2 and vaccines reported the occurrence of autoimmune thyroiditis and autoimmune insulitis, but these events were infrequent.69  Perhaps as cancer vaccines become more potent as they can be in animal models, we might see a concomitant increase in autoimmune sequelae.

Table 4

Autoimmunity associated with cancer vaccines

VaccineMalignancy (species)Autoimmune eventsReference(s)
TRP-1 encoded by virus Melanoma (mouse) Vitiligo 120,121  
CD20 + adjuvant Lymphoma (mouse) Depletion of normal B cells 122  
Her-2 encoded by plasmids Breast cancer–expressing Her-2 (mouse) Impaired lactation, accelerated involution of mammary gland 123,124  
DCs (peptide pulsed) CD40 and IL-2 (fibroblasts) Lymphoblastic leukemia (mouse) Systemic autoimmunity, immunity against fibroblasts, hepatomegaly, splenomegaly, fur loss, cachexia, 125  
DCs (peptide-pulsed) β-gal and LCMV model antigens EL-4 thymoma (mouse) Diabetes, myocarditis 126  
Pulsed DCs or tumor cells and either GM-CSF and adjuvant or GM-CSF, adjuvant, and peptides Melanoma (human) Vitiligo, diabetes, thyroiditis, ocular toxicity 69  
VaccineMalignancy (species)Autoimmune eventsReference(s)
TRP-1 encoded by virus Melanoma (mouse) Vitiligo 120,121  
CD20 + adjuvant Lymphoma (mouse) Depletion of normal B cells 122  
Her-2 encoded by plasmids Breast cancer–expressing Her-2 (mouse) Impaired lactation, accelerated involution of mammary gland 123,124  
DCs (peptide pulsed) CD40 and IL-2 (fibroblasts) Lymphoblastic leukemia (mouse) Systemic autoimmunity, immunity against fibroblasts, hepatomegaly, splenomegaly, fur loss, cachexia, 125  
DCs (peptide-pulsed) β-gal and LCMV model antigens EL-4 thymoma (mouse) Diabetes, myocarditis 126  
Pulsed DCs or tumor cells and either GM-CSF and adjuvant or GM-CSF, adjuvant, and peptides Melanoma (human) Vitiligo, diabetes, thyroiditis, ocular toxicity 69  

DCs indicates dendritic cells; LCMV, lymphocytic choriomeningitis virus; and GM-CSF, granulocyte-macrophage colony-stimulating factor.

Although immunotherapy holds great promise for the treatment of a range of malignancies, many of these therapies can be associated with autoimmunity against normal self-tissues (Figure 1). Mechanisms involved in immune-mediated damage of normal tissues are varied, and a greater understanding of these mechanisms will enable enhanced and more specific forms of immunotherapy for cancer.

The most obvious means of toxicity results from expression of TAAs on normal tissue (Figure 2). Thus, antibodies raised against these antigens can react against normal cells either by inducing complement-mediated lysis or facilitating ADCC by innate leukocytes. Adoptively transferred T cells specific for TAAs can likewise react directly against normal tissues expressing those antigens as part of their normal proteome. However, toxicity can also result from de novo induction of immune responses against other antigens on normal tissues. For example, inhibition of CTLA-4 can dysregulate normal lymphocyte homeostasis, leading to expansion of self-reactive T cells able to respond against normal tissues that do not express TAAs.70  Similarly, immunotherapy can lead to epitope spreading in which immune responses can be raised against additional molecular targets, including those expressed on normal tissues.71 

Figure 2

Mechanisms of immune toxicity associated with immunotherapy. (1) The delivery of exogenous antibody specific for TAA expressed on both tumor and normal tissue can result in damage to normal tissue mediated by complement or ADCC mediated by innate immune cells, such as macrophages. (2) Similarly, transfer of TAA-specific T cells can lead to destruction of normal tissue if the TAA is also expressed on cells of normal tissue. (3) Alternate ways of inducing immune reactivity toward normal tissue include dysregulation of normal immune homeostasis using anti–CTLA-4, resulting in expansion of self-reactive T cells. (4) It is also possible that induction of immunity to tumor cells could lead to epitope spreading whereby T cells reactive with self-antigens are generated.

Figure 2

Mechanisms of immune toxicity associated with immunotherapy. (1) The delivery of exogenous antibody specific for TAA expressed on both tumor and normal tissue can result in damage to normal tissue mediated by complement or ADCC mediated by innate immune cells, such as macrophages. (2) Similarly, transfer of TAA-specific T cells can lead to destruction of normal tissue if the TAA is also expressed on cells of normal tissue. (3) Alternate ways of inducing immune reactivity toward normal tissue include dysregulation of normal immune homeostasis using anti–CTLA-4, resulting in expansion of self-reactive T cells. (4) It is also possible that induction of immunity to tumor cells could lead to epitope spreading whereby T cells reactive with self-antigens are generated.

Close modal

As our knowledge of the human proteome and the cancer genome increases, opportunities are arising to refine the choice of antigens for immunotherapy. Next-generation genome sequencing allows cataloging of all the mutations within any given tumor cell.72  This could allow the rapid identification of truly tumor-unique antigens and greater personalization of vaccine and adoptive immunotherapies in the near future. Already anti-idiotype vaccines are showing promise for the specific targeting of malignant B-cell lymphomas.73,74 

Methods to reduce immune-mediated toxicity in adoptive immunotherapy may involve identifying multiple antigens that together constitute a tumor signature or “barcode.”75  Gene constructs encoding several antigen receptors can be developed where the threshold for cytotoxic activity is only reached if the complete “barcode” is recognized. Alternatively, constructs can be designed which shut down T-cell activity if a barcode indicative of normal healthy tissue is encountered.

In the case of cytokine therapy, systemic toxicities and the deregulation of immune responses may also be reduced through more targeted delivery of cytokines to the tumor site, rather than systemically.76 

In antibody therapy, it is useful to consider that not all antibodies for a specific target molecule are equal in their immune side effects, as seen using TGN1412, a monoclonal antibody specific for the T-cell costimulatory molecule CD28. TGN1412 was thought to hold promise as a treatment for cancer and rheumatoid arthritis, but a safety trial performed in 6 healthy volunteers demonstrated severe toxicity, including hypotension and respiratory distress.77  Toxicity was thought to be the result of high levels of serum cytokines in response to CD28 ligation occurring within hours of antibody administration. TGN1412 differed from other anti-CD28 antibodies in that it had superagonist qualities, not possessed by all anti-CD28 antibodies. Similarly, individual anti-CTLA-4 antibodies can differ in their capacity to induce autoimmunity, as demonstrated in mice engineered to express human CTLA-4. In this mouse model, several antibodies varied in their relative abilities to induce autoimmunity and protection against tumor.78  An antibody was identified that produced the strongest antitumor activity and the least autoimmunity. Thus, it may be possible to separate antitumor activity from autoimmunity by antibody selection.

Selective down-regulation of autoimmunity may also be possible using immunosuppressants. The use of corticosteroids to counteract severe immune-mediated toxicity has surprisingly indicated that steroid treatment may not always signal the end of an immunotherapy's beneficial impact, with tumor responses being maintained after resolution of autoimmunity using steroids.31,79  A further consideration is whether it is possible to kinetically separate antitumor immunity from autoimmunity. Regulatory mechanisms on healthy tissues may allow them to withstand immune toxicity for a period of time.

Immunotherapy is an exciting and increasingly effective treatment option for cancer. However, it is becoming increasingly clear that cancer immunotherapy is a balancing act between antitumor immunity and immune toxicity. The association between immune toxicity and increased antitumor effects after immunotherapy highlights the need for strategies that can mitigate the risk of these toxicities during immunotherapy while preserving activity against malignancy.

This work was supported by the National Health and Medical Research Council of Australia, Cancer Council of Victoria, and the Susan G. Komen Breast Cancer Foundation. M.H.K. is supported by a Senior Research Fellowship from the National Health and Medical Research Council of Australia. P.K.D. is supported by a National Health and Medical Research Council of Australia Career Development Award. S.M.A. is supported by a Cancer Council of Victoria Postgraduate Cancer Research Scholarship.

Contribution: S.M.A., C.P.M.D., J.A.W., D.S.R., P.K.D., and M.H.K. researched and wrote the paper; and R.P.J. assisted in writing the adoptive immunotherapy section and critically reviewed the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Michael H. Kershaw, Cancer Immunology Research Program, Peter MacCallum Cancer Centre, St Andrews Place, Melbourne, Victoria, Australia 3002; e-mail: michael.kershaw@petermac.org.

1
Banchereau
 
J
Palucka
 
AK
Dendritic cells as therapeutic vaccines against cancer.
Nat Rev Immunol
2005
, vol. 
5
 
4
(pg. 
296
-
306
)
2
Smyth
 
MJ
Cretney
 
E
Kershaw
 
MH
Hayakawa
 
Y
Cytokines in cancer immunity and immunotherapy.
Immunol Rev
2004
, vol. 
202
 (pg. 
275
-
293
)
3
Weiner
 
LM
Dhodapkar
 
MV
Ferrone
 
S
Monoclonal antibodies for cancer immunotherapy.
Lancet
2009
, vol. 
373
 
9668
(pg. 
1033
-
1040
)
4
Rosenberg
 
SA
Dudley
 
ME
Adoptive cell therapy for the treatment of patients with metastatic melanoma.
Curr Opin Immunol
2009
, vol. 
21
 
2
(pg. 
233
-
240
)
5
Bollard
 
CM
Aguilar
 
L
Straathof
 
KC
, et al. 
Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin's disease.
J Exp Med
2004
, vol. 
200
 
12
(pg. 
1623
-
1633
)
6
Grimm
 
EA
Robb
 
RJ
Roth
 
JA
, et al. 
Lymphokine-activated killer cell phenomenon: III. Evidence that IL-2 is sufficient for direct activation of peripheral blood lymphocytes into lymphokine-activated killer cells.
J Exp Med
1983
, vol. 
158
 
4
(pg. 
1356
-
1361
)
7
Mitchison
 
NA
Studies on the immunological response to foreign tumor transplants in the mouse: I. The role of lymph node cells in conferring immunity by adoptive transfer.
J Exp Med
1955
, vol. 
102
 
2
(pg. 
157
-
177
)
8
Overwijk
 
WW
Theoret
 
MR
Finkelstein
 
SE
, et al. 
Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells.
J Exp Med
2003
, vol. 
198
 
4
(pg. 
569
-
580
)
9
Dudley
 
ME
Yang
 
JC
Sherry
 
R
, et al. 
Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens.
J Clin Oncol
2008
, vol. 
26
 
32
(pg. 
5233
-
5239
)
10
Johnson
 
LA
Morgan
 
RA
Dudley
 
ME
, et al. 
Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen.
Blood
2009
, vol. 
114
 
3
(pg. 
535
-
546
)
11
Bendle
 
GM
Linnemann
 
C
Hooijkaas
 
AI
, et al. 
Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy.
Nat Med
2010
, vol. 
16
 
5
(pg. 
565
-
570
)
12
Rosenberg
 
SA
Of mice, not men: no evidence for graft-versus-host disease in humans receiving T-cell receptor-transduced autologous T cells.
Mol Ther
2010
, vol. 
18
 
10
(pg. 
1744
-
1745
)
13
Westwood
 
JA
Kershaw
 
MH
Genetic redirection of T cells for cancer therapy.
J Leukoc Biol
2010
, vol. 
87
 
5
(pg. 
791
-
803
)
14
Lamers
 
CH
Sleijfer
 
S
Vulto
 
AG
, et al. 
Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience.
J Clin Oncol
2006
, vol. 
24
 
13
(pg. 
e20
-
e22
)
15
Kochenderfer
 
JN
Wilson
 
WH
Janik
 
JE
, et al. 
Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19.
Blood
2010
, vol. 
116
 
20
(pg. 
4099
-
4102
)
16
Brentjens
 
R
Yeh
 
R
Bernal
 
Y
Riviere
 
I
Sadelain
 
M
Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial.
Mol Ther
2010
, vol. 
18
 
4
(pg. 
666
-
668
)
17
Morgan
 
RA
Yang
 
JC
Kitano
 
M
Dudley
 
ME
Laurencot
 
CM
Rosenberg
 
SA
Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2.
Mol Ther
2010
, vol. 
18
 
4
(pg. 
843
-
851
)
18
Bernhard
 
H
Neudorfer
 
J
Gebhard
 
K
, et al. 
Adoptive transfer of autologous, HER2-specific, cytotoxic T lymphocytes for the treatment of HER2-overexpressing breast cancer.
Cancer Immunol Immunother
2008
, vol. 
57
 
2
(pg. 
271
-
280
)
19
Parkhurst
 
MR
Yang
 
JC
Langan
 
RC
, et al. 
T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis.
Mol Ther
2011
, vol. 
19
 
3
(pg. 
620
-
626
)
20
Chen
 
YT
Scanlan
 
MJ
Sahin
 
U
, et al. 
A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
5
(pg. 
1914
-
1918
)
21
Robbins
 
PF
Morgan
 
RA
Feldman
 
SA
, et al. 
Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1.
J Clin Oncol
2011
, vol. 
29
 
7
(pg. 
917
-
924
)
22
Pule
 
MA
Savoldo
 
B
Myers
 
GD
, et al. 
Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma.
Nat Med
2008
, vol. 
14
 
11
(pg. 
1264
-
1270
)
23
Weiner
 
LM
Surana
 
R
Wang
 
S
Monoclonal antibodies: versatile platforms for cancer immunotherapy.
Nat Rev Immunol
2010
, vol. 
10
 
5
(pg. 
317
-
327
)
24
Roberts
 
WK
Darnell
 
RB
Neuroimmunology of the paraneoplastic neurological degenerations.
Curr Opin Immunol
2004
, vol. 
16
 
5
(pg. 
616
-
622
)
25
Diaz
 
JP
Tew
 
WP
Zivanovic
 
O
, et al. 
Incidence and management of bevacizumab-associated gastrointestinal perforations in patients with recurrent ovarian carcinoma.
Gynecol Oncol
2010
, vol. 
116
 
3
(pg. 
335
-
339
)
26
Lin
 
WL
Lin
 
WC
Yang
 
JY
, et al. 
Fatal toxic epidermal necrolysis associated with cetuximab in a patient with colon cancer.
J Clin Oncol
2008
, vol. 
26
 
16
(pg. 
2779
-
2780
)
27
Slamon
 
DJ
Leyland-Jones
 
B
Shak
 
S
, et al. 
Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.
N Engl J Med
2001
, vol. 
344
 
11
(pg. 
783
-
792
)
28
Phan
 
GQ
Yang
 
JC
Sherry
 
RM
, et al. 
Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
14
(pg. 
8372
-
8377
)
29
Bashey
 
A
Medina
 
B
Corringham
 
S
, et al. 
CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation.
Blood
2009
, vol. 
113
 
7
(pg. 
1581
-
1588
)
30
Beck
 
KE
Blansfield
 
JA
Tran
 
KQ
, et al. 
Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4.
J Clin Oncol
2006
, vol. 
24
 
15
(pg. 
2283
-
2289
)
31
Wolchok
 
JD
Neyns
 
B
Linette
 
G
, et al. 
Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study.
Lancet Oncol
2010
, vol. 
11
 
2
(pg. 
155
-
164
)
32
Klein
 
O
Ebert
 
LM
Nicholaou
 
T
, et al. 
Melan-A-specific cytotoxic T cells are associated with tumor regression and autoimmunity following treatment with anti-CTLA-4.
Clin Cancer Res
2009
, vol. 
15
 
7
(pg. 
2507
-
2513
)
33
Hodi
 
FS
O'Day
 
SJ
McDermott
 
DF
, et al. 
Improved survival with ipilimumab in patients with metastatic melanoma.
N Engl J Med
2010
, vol. 
363
 
8
(pg. 
711
-
723
)
34
Brahmer
 
JR
Drake
 
CG
Wollner
 
I
, et al. 
Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates.
J Clin Oncol
2010
, vol. 
28
 
19
(pg. 
3167
-
3175
)
35
Linenberger
 
ML
CD33-directed therapy with gemtuzumab ozogamicin in acute myeloid leukemia: progress in understanding cytotoxicity and potential mechanisms of drug resistance.
Leukemia
2005
, vol. 
19
 
2
(pg. 
176
-
182
)
36
Larson
 
RA
Sievers
 
EL
Stadtmauer
 
EA
, et al. 
Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence.
Cancer
2005
, vol. 
104
 
7
(pg. 
1442
-
1452
)
37
Sievers
 
EL
Larson
 
RA
Stadtmauer
 
EA
, et al. 
Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse.
J Clin Oncol
2001
, vol. 
19
 
13
(pg. 
3244
-
3254
)
38
McKoy
 
JM
Angelotta
 
C
Bennett
 
CL
, et al. 
Gemtuzumab ozogamicin-associated sinusoidal obstructive syndrome (SOS): an overview from the research on adverse drug events and reports (RADAR) project.
Leuk Res
2007
, vol. 
31
 
5
(pg. 
599
-
604
)
39
Rajvanshi
 
P
Shulman
 
HM
Sievers
 
EL
McDonald
 
GB
Hepatic sinusoidal obstruction after gemtuzumab ozogamicin (Mylotarg) therapy.
Blood
2002
, vol. 
99
 
7
(pg. 
2310
-
2314
)
40
Burnett
 
AK
Hills
 
RK
Milligan
 
D
, et al. 
Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 Trial.
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
369
-
377
)
41
Hale
 
G
The CD52 antigen and development of the CAMPATH antibodies.
Cytotherapy
2001
, vol. 
3
 
3
(pg. 
137
-
143
)
42
Lin
 
TS
Donohue
 
KA
Byrd
 
JC
, et al. 
Consolidation therapy with subcutaneous alemtuzumab after fludarabine and rituximab induction therapy for previously untreated chronic lymphocytic leukemia: final analysis of CALGB 10101.
J Clin Oncol
2010
, vol. 
28
 
29
(pg. 
4500
-
4506
)
43
Jiang
 
L
Yuan
 
CM
Hubacheck
 
J
, et al. 
Variable CD52 expression in mature T cell and NK cell malignancies: implications for alemtuzumab therapy.
Br J Haematol
2009
, vol. 
145
 
2
(pg. 
173
-
179
)
44
Wendtner
 
CM
Ritgen
 
M
Schweighofer
 
CD
, et al. 
Consolidation with alemtuzumab in patients with chronic lymphocytic leukemia (CLL) in first remission: experience on safety and efficacy within a randomized multicenter phase III trial of the German CLL Study Group (GCLLSG).
Leukemia
2004
, vol. 
18
 
6
(pg. 
1093
-
1101
)
45
Maloney
 
DG
Smith
 
B
Rose
 
A
Rituximab: mechanism of action and resistance.
Semin Oncol
2002
, vol. 
29
 
1 suppl 2
(pg. 
2
-
9
)
46
Feugier
 
P
Van Hoof
 
A
Sebban
 
C
, et al. 
Long-term results of the R-CHOP study in the treatment of elderly patients with diffuse large B-cell lymphoma: a study by the Groupe d'Etude des Lymphomes de l'Adulte.
J Clin Oncol
2005
, vol. 
23
 
18
(pg. 
4117
-
4126
)
47
Schulz
 
H
Bohlius
 
JF
Trelle
 
S
, et al. 
Immunochemotherapy with rituximab and overall survival in patients with indolent or mantle cell lymphoma: a systematic review and meta-analysis.
J Natl Cancer Inst
2007
, vol. 
99
 
9
(pg. 
706
-
714
)
48
Garcia-Rodriguez
 
MJ
Canales
 
MA
Hernandez-Maraver
 
D
Hernandez-Navarro
 
F
Late reactivation of resolved hepatitis B virus infection: an increasing complication post rituximab-based regimens treatment?
Am J Hematol
2008
, vol. 
83
 
8
(pg. 
673
-
675
)
49
Hirokawa
 
M
Kawabata
 
Y
Fujishima
 
N
Yoshioka
 
T
Sawada
 
K
Prolonged reactivation of cytomegalovirus infection following successful rituximab therapy for Epstein-Barr virus-associated posttransplantation lymphoproliferative disorder.
Int J Hematol
2007
, vol. 
86
 
3
(pg. 
291
-
292
)
50
Grant
 
C
Wilson
 
WH
Dunleavy
 
K
Neutropenia associated with rituximab therapy.
Curr Opin Hematol
2011
, vol. 
18
 
1
(pg. 
49
-
54
)
51
Steegmann
 
JL
Requena
 
MJ
Martin-Regueira
 
P
, et al. 
High incidence of autoimmune alterations in chronic myeloid leukemia patients treated with interferon-alpha.
Am J Hematol
2003
, vol. 
72
 
3
(pg. 
170
-
176
)
52
Gogas
 
H
Ioannovich
 
J
Dafni
 
U
, et al. 
Prognostic significance of autoimmunity during treatment of melanoma with interferon.
N Engl J Med
2006
, vol. 
354
 
7
(pg. 
709
-
718
)
53
Dunn
 
GP
Bruce
 
AT
Sheehan
 
KC
, et al. 
A critical function for type I interferons in cancer immunoediting.
Nat Immunol
2005
, vol. 
6
 
7
(pg. 
722
-
729
)
54
Carson
 
WE
Interferon-alpha-induced activation of signal transducer and activator of transcription proteins in malignant melanoma.
Clin Cancer Res
1998
, vol. 
4
 
9
(pg. 
2219
-
2228
)
55
Afkarian
 
M
Sedy
 
JR
Yang
 
J
, et al. 
T-bet is a STAT1-induced regulator of IL-12R expression in naive CD4+ T cells.
Nat Immunol
2002
, vol. 
3
 
6
(pg. 
549
-
557
)
56
Dunn
 
IS
Haggerty
 
TJ
Kono
 
M
, et al. 
Enhancement of human melanoma antigen expression by IFN-beta.
J Immunol
2007
, vol. 
179
 
4
(pg. 
2134
-
2142
)
57
Marrack
 
P
Kappler
 
J
Mitchell
 
T
Type I interferons keep activated T cells alive.
J Exp Med
1999
, vol. 
189
 
3
(pg. 
521
-
530
)
58
Tovey
 
MG
Lallemand
 
C
Thyphronitis
 
G
Adjuvant activity of type I interferons.
Biol Chem
2008
, vol. 
389
 
5
(pg. 
541
-
545
)
59
Atkins
 
MB
Lotze
 
MT
Dutcher
 
JP
, et al. 
High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993.
J Clin Oncol
1999
, vol. 
17
 
7
(pg. 
2105
-
2116
)
60
Klapper
 
JA
Downey
 
SG
Smith
 
FO
, et al. 
High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma: a retrospective analysis of response and survival in patients treated in the surgery branch at the National Cancer Institute between 1986 and 2006.
Cancer
2008
, vol. 
113
 
2
(pg. 
293
-
301
)
61
Schwartzentruber
 
DJ
Guidelines for the safe administration of high-dose interleukin-2.
J Immunother
2001
, vol. 
24
 
4
(pg. 
287
-
293
)
62
Fraenkel
 
PG
Rutkove
 
SB
Matheson
 
JK
, et al. 
Induction of myasthenia gravis, myositis, and insulin-dependent diabetes mellitus by high-dose interleukin-2 in a patient with renal cell cancer.
J Immunother
2002
, vol. 
25
 
4
(pg. 
373
-
378
)
63
Franzke
 
A
Peest
 
D
Probst-Kepper
 
M
, et al. 
Autoimmunity resulting from cytokine treatment predicts long-term survival in patients with metastatic renal cell cancer.
J Clin Oncol
1999
, vol. 
17
 
2
(pg. 
529
-
533
)
64
Weijl
 
NI
Van der Harst
 
D
Brand
 
A
, et al. 
Hypothyroidism during immunotherapy with interleukin-2 is associated with antithyroid antibodies and response to treatment.
J Clin Oncol
1993
, vol. 
11
 
7
(pg. 
1376
-
1383
)
65
Soni
 
N
Meropol
 
NJ
Porter
 
M
Caligiuri
 
MA
Diabetes mellitus induced by low-dose interleukin-2.
Cancer Immunol Immunother
1996
, vol. 
43
 
1
(pg. 
59
-
62
)
66
Abdel-Naser
 
MB
Kruger-Krasagakes
 
S
Krasagakis
 
K
Gollnick
 
H
Abdel-Fattah
 
A
Orfanos
 
CE
Further evidence for involvement of both cell mediated and humoral immunity in generalized vitiligo.
Pigment Cell Res
1994
, vol. 
7
 
1
(pg. 
1
-
8
)
67
Rosenberg
 
SA
White
 
DE
Vitiligo in patients with melanoma: normal tissue antigens can be targets for cancer immunotherapy.
J Immunother Emphasis Tumor Immunol
1996
, vol. 
19
 
1
(pg. 
81
-
84
)
68
Rosenberg
 
SA
Yang
 
JC
Restifo
 
NP
Cancer immunotherapy: moving beyond current vaccines.
Nat Med
2004
, vol. 
10
 
9
(pg. 
909
-
915
)
69
Chianese-Bullock
 
KA
Woodson
 
EM
Tao
 
H
, et al. 
Autoimmune toxicities associated with the administration of antitumor vaccines and low-dose interleukin-2.
J Immunother
2005
, vol. 
28
 
4
(pg. 
412
-
419
)
70
Jain
 
N
Nguyen
 
H
Chambers
 
C
Kang
 
J
Dual function of CTLA-4 in regulatory T cells and conventional T cells to prevent multiorgan autoimmunity.
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
4
(pg. 
1524
-
1528
)
71
Corbière
 
V
Chapiro
 
J
Stroobant
 
V
, et al. 
Antigen spreading contributes to MAGE vaccination-induced regression of melanoma metastases.
Cancer Res
2011
, vol. 
71
 
4
(pg. 
1253
-
1262
)
72
Lee
 
W
Jiang
 
Z
Liu
 
J
, et al. 
The mutation spectrum revealed by paired genome sequences from a lung cancer patient.
Nature
2010
, vol. 
465
 
7297
(pg. 
473
-
477
)
73
Timmerman
 
JM
Singh
 
G
Hermanson
 
G
, et al. 
Immunogenicity of a plasmid DNA vaccine encoding chimeric idiotype in patients with B-cell lymphoma.
Cancer Res
2002
, vol. 
62
 
20
(pg. 
5845
-
5852
)
74
Mahaseth
 
H
Brody
 
JD
Sinha
 
R
Shenoy
 
PJ
Flowers
 
CR
Idiotype vaccine strategies for treatment of follicular lymphoma.
Future Oncol
2010
, vol. 
7
 
1
(pg. 
111
-
122
)
75
da Cunha
 
JP
Galante
 
PA
de Souza
 
JE
, et al. 
Bioinformatics construction of the human cell surfaceome.
Proc Natl Acad Sci U S A
2009
, vol. 
106
 
39
(pg. 
16752
-
16757
)
76
Kaspar
 
M
Trachsel
 
E
Neri
 
D
The antibody-mediated targeted delivery of interleukin-15 and GM-CSF to the tumor neovasculature inhibits tumor growth and metastasis.
Cancer Res
2007
, vol. 
67
 
10
(pg. 
4940
-
4948
)
77
Suntharalingam
 
G
Perry
 
MR
Ward
 
S
, et al. 
Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412.
N Engl J Med
2006
, vol. 
355
 
10
(pg. 
1018
-
1028
)
78
Lute
 
KD
May
 
KF
Lu
 
P
, et al. 
Human CTLA4 knock-in mice unravel the quantitative link between tumor immunity and autoimmunity induced by anti-CTLA-4 antibodies.
Blood
2005
, vol. 
106
 
9
(pg. 
3127
-
3133
)
79
Attia
 
P
Phan
 
G
Maker
 
A
, et al. 
Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4.
J Clin Oncol
2005
, vol. 
23
 (pg. 
6043
-
6053
)
80
Pellkofer
 
H
Schubart
 
AS
Hoftberger
 
R
, et al. 
Modelling paraneoplastic CNS disease: T-cells specific for the onconeuronal antigen PNMA1 mediate autoimmune encephalomyelitis in the rat.
Brain
2004
, vol. 
127
 
Pt 8
(pg. 
1822
-
1830
)
81
Maeda
 
A
Maeda
 
T
Ohguro
 
H
Palczewski
 
K
Sato
 
N
Vaccination with recoverin, a cancer-associated retinopathy antigen, induces autoimmune retinal dysfunction and tumor cell regression in mice.
Eur J Immunol
2002
, vol. 
32
 
8
(pg. 
2300
-
2307
)
82
Palmer
 
DC
Chan
 
CC
Gattinoni
 
L
, et al. 
Effective tumor treatment targeting a melanoma/melanocyte-associated antigen triggers severe ocular autoimmunity.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
23
(pg. 
8061
-
8066
)
83
Quezada
 
SA
Simpson
 
TR
Peggs
 
KS
, et al. 
Tumor-reactive CD4(+) T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts.
J Exp Med
2010
, vol. 
207
 
3
(pg. 
637
-
650
)
84
Xie
 
Y
Akpinarli
 
A
Maris
 
C
, et al. 
Naive tumor-specific CD4(+) T cells differentiated in vivo eradicate established melanoma.
J Exp Med
2010
, vol. 
207
 
3
(pg. 
651
-
667
)
85
Bos
 
R
van Duikeren
 
S
Morreau
 
H
, et al. 
Balancing between antitumor efficacy and autoimmune pathology in T-cell-mediated targeting of carcinoembryonic antigen.
Cancer Res
2008
, vol. 
68
 
20
(pg. 
8446
-
8455
)
86
Ugel
 
S
Scarselli
 
E
Iezzi
 
M
, et al. 
Autoimmune B-cell lymphopenia after successful adoptive therapy with telomerase-specific T lymphocytes.
Blood
2010
, vol. 
115
 
7
(pg. 
1374
-
1384
)
87
Cheadle
 
EJ
Hawkins
 
RE
Batha
 
H
O'Neill
 
AL
Dovedi
 
SJ
Gilham
 
DE
Natural expression of the CD19 antigen impacts the long-term engraftment but not antitumor activity of CD19-specific engineered T cells.
J Immunol
2010
, vol. 
184
 
4
(pg. 
1885
-
1896
)
88
Kochenderfer
 
JN
Yu
 
Z
Frasheri
 
D
Restifo
 
NP
Rosenberg
 
SA
Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells.
Blood
2010
, vol. 
116
 
19
(pg. 
3875
-
3886
)
89
Chinnasamy
 
D
Yu
 
Z
Theoret
 
MR
, et al. 
Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenic tumors in mice.
J Clin Invest
2010
, vol. 
120
 
11
(pg. 
3953
-
3968
)
90
Rosenberg
 
SA
Packard
 
BS
Aebersold
 
PM
, et al. 
Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma: a preliminary report.
N Engl J Med
1988
, vol. 
319
 
25
(pg. 
1676
-
1680
)
91
Ridolfi
 
LRR
Riccobon
 
A
De Paola
 
F
, et al. 
Adjuvant adoptive immunotherapy in patients with stage III and resected stage IV melanoma: a pilot study.
J Immunother
2003
, vol. 
26
 
2
(pg. 
156
-
162
)
92
Yeh
 
S
Karne
 
NK
Kerkar
 
SP
, et al. 
Ocular and systemic autoimmunity after successful tumor-infiltrating lymphocyte immunotherapy for recurrent, metastatic melanoma.
Ophthalmology
2009
, vol. 
116
 
5
(pg. 
981
-
989
)
93
Dudley
 
ME
Wunderlich
 
JR
Robbins
 
PF
, et al. 
Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes.
Science
2002
, vol. 
298
 
5594
(pg. 
850
-
854
)
94
Yee
 
C
Thompson
 
JA
Roche
 
P
, et al. 
Melanocyte destruction after antigen-specific immunotherapy of melanoma: direct evidence of T cell-mediated vitiligo.
J Exp Med
2000
, vol. 
192
 
11
(pg. 
1637
-
1644
)
95
Camacho
 
LH
Antonia
 
S
Sosman
 
J
, et al. 
Phase I/II trial of tremelimumab in patients with metastatic melanoma.
J Clin Oncol
2009
, vol. 
27
 
7
(pg. 
1075
-
1081
)
96
Attia
 
P
Phan
 
GQ
Maker
 
AV
, et al. 
Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4.
J Clin Oncol
2005
, vol. 
23
 
25
(pg. 
6043
-
6053
)
97
Hodi
 
FS
Mihm
 
MC
Soiffer
 
RJ
, et al. 
Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
8
(pg. 
4712
-
4717
)
98
Robinson
 
MR
Chan
 
CC
Yang
 
JC
, et al. 
Cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma: a new cause of uveitis.
J Immunother
2004
, vol. 
27
 
6
(pg. 
478
-
479
)
99
Sanderson
 
K
Scotland
 
R
Lee
 
P
, et al. 
Autoimmunity in a phase I trial of a fully human anti-cytotoxic T-lymphocyte antigen-4 monoclonal antibody with multiple melanoma peptides and Montanide ISA 51 for patients with resected stages III and IV melanoma.
J Clin Oncol
2005
, vol. 
23
 
4
(pg. 
741
-
750
)
100
van Elsas
 
A
Hurwitz
 
AA
Allison
 
JP
Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation.
J Exp Med
1999
, vol. 
190
 
3
(pg. 
355
-
366
)
101
Gregor
 
PD
Wolchok
 
JD
Ferrone
 
CR
, et al. 
CTLA-4 blockade in combination with xenogeneic DNA vaccines enhances T-cell responses, tumor immunity and autoimmunity to self antigens in animal and cellular model systems.
Vaccine
2004
, vol. 
22
 
13
(pg. 
1700
-
1708
)
102
van Elsas
 
A
Sutmuller
 
RPM
Hurwitz
 
AA
, et al. 
Elucidating the autoimmune and antitumor effector mechanisms of a treatment based on cytotoxic T lymphocyte antigen-4 blockade in combination with a B16 melanoma vaccine: comparison of prophylaxis and therapy.
J Exp Med
2001
, vol. 
194
 
4
(pg. 
481
-
490
)
103
Hurwitz
 
AA
Foster
 
BA
Kwon
 
ED
, et al. 
Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade.
Cancer Res
2000
, vol. 
60
 
9
(pg. 
2444
-
2448
)
104
Moreton
 
P
Hillmen
 
P
Alemtuzumab therapy in B-cell lymphoproliferative disorders.
Semin Oncol
2003
, vol. 
30
 
4
(pg. 
493
-
501
)
105
Foran
 
JM
Rohatiner
 
AZS
Cunningham
 
D
, et al. 
European phase II study of rituximab (chimeric anti-CD20 monoclonal antibody) for patients with newly diagnosed mantle-cell lymphoma and previously treated mantle-cell lymphoma, immunocytoma, and small B-cell lymphocytic lymphoma.
J Clin Oncol
2000
, vol. 
18
 
2
(pg. 
317
-
324
)
106
Suzan
 
F
Ammor
 
M
Ribrag
 
V
Fatal reactivation of cytomegalovirus infection after use of rituximab for a post-transplantation lymphoproliferative disorder.
N Engl J Med
2001
, vol. 
345
 
13
pg. 
1000
 
107
Herishanu
 
Y
Trestman
 
S
Kirgner
 
I
Rachmani
 
R
Naparstek
 
E
Autoimmune thrombocytopenia in chronic myeloid leukemia treated with interferon-alpha: differential diagnosis and possible pathogenesis.
Leuk Lymphoma
2003
, vol. 
44
 
12
(pg. 
2103
-
2108
)
108
Ladoyanni
 
E
Nambi
 
R
Psoriasis exacerbated by interferon-alpha in a patient with chronic myeloid leukemia.
J Drugs Dermatol
2005
, vol. 
4
 
2
(pg. 
221
-
222
)
109
Ozet
 
A
Ozet
 
G
Caliskaner
 
Z
Komurcu
 
S
Ozturk
 
B
Development of hyperthyroidism during long term interferon therapy in a patient with chronic myelogenous leukemia: case report.
Endocr J
1997
, vol. 
44
 
5
(pg. 
715
-
717
)
110
Tannir
 
NM
Talpaz
 
M
Ghazal
 
H
Proothi
 
S
Kantarjian
 
HM
Acute pancreatitis associated with interferon alpha therapy for chronic myelogenous leukemia.
Leuk Lymphoma
2000
, vol. 
39
 
5
(pg. 
647
-
650
)
111
Wolfe
 
JT
Singh
 
A
Lessin
 
SR
Jaworsky
 
C
Rook
 
AH
De novo development of psoriatic plaques in patients receiving interferon alfa for treatment of erythrodermic cutaneous T-cell lymphoma.
J Am Acad Dermatol
1995
, vol. 
32
 
5
(pg. 
887
-
893
)
112
Ronnblom
 
LE
Alm
 
GV
Oberg
 
KE
Autoimmunity after alpha-interferon therapy for malignant carcinoid tumors.
Ann Intern Med
1991
, vol. 
115
 
3
(pg. 
178
-
183
)
113
Aslan
 
M
Nazlıgul
 
Y
Aksoy
 
N
Yılmaz
 
N
A case of hyperthyroidy developing in “pegylated-interferon” therapy.
Dig Dis Sci
2007
, vol. 
52
 
5
pg. 
1194
 
114
Harris
 
J
Bines
 
S
Das Gupta
 
T
Therapy of disseminated malignant melanoma with recombinant alpha 2b-interferon and piroxicam: clinical results with a report of an unusual response-associated feature (vitiligo) and unusual toxicity (diffuse pulmonary interstitial fibrosis).
Med Pediatr Oncol
1994
, vol. 
22
 
2
(pg. 
103
-
106
)
115
Slingluff
 
C
Petroni
 
G
Yamshchikov
 
G
, et al. 
Clinical and immunologic results of a randomized phase II trial of vaccination using four melanoma peptides either administered in granulocyte-macrophage colony-stimulating factor in adjuvant or pulsed on dendritic cells.
J Clin Oncol
2003
, vol. 
21
 
21
(pg. 
4016
-
4026
)
116
Esteva-Lorenzo
 
FJ
Janik
 
JE
Fenton
 
RG
Emslie-Smith
 
A
Engel
 
AG
Longo
 
DL
Myositis associated with interleukin-2 therapy in a patient with metastatic renal cell carcinoma.
Cancer
1995
, vol. 
76
 
7
(pg. 
1219
-
1223
)
117
Soni
 
N
Meropol
 
NJ
Porter
 
M
Caligiuri
 
MA
Diabetes mellitus induced by low-dose interleukin-2.
Cancer Immunol Immunother
1996
, vol. 
43
 
1
(pg. 
59
-
62
)
118
Sauter
 
NP
Atkins
 
MB
Mier
 
JW
Lechan
 
RM
Transient thyrotoxicosis and persistent hypothyroidism due to acute autoimmune thyroiditis after interleukin-2 and interferon-alpha therapy for metastatic carcinoma: a case report.
Am J Med
1992
, vol. 
92
 
4
pg. 
441
 
119
Scalzo
 
SGA
Boccoli
 
G
Masciulli
 
R
, et al. 
Primary hypothyroidism associated with interleukin-2 and interferon alpha-2 therapy of melanoma and renal carcinoma.
Eur J Cancer
1990
, vol. 
26
 
11
(pg. 
1152
-
1156
)
120
Weber
 
LW
Bowne
 
WB
Wolchok
 
JD
, et al. 
Tumor immunity and autoimmunity induced by immunization with homologous DNA.
J Clin Invest
1998
, vol. 
102
 
6
(pg. 
1258
-
1264
)
121
Overwijk
 
WW
Lee
 
DS
Surman
 
DR
, et al. 
Vaccination with a recombinant vaccinia virus encoding a “self” antigen induces autoimmune vitiligo and tumor cell destruction in mice: requirement for CD4(+) T lymphocytes.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 
6
(pg. 
2982
-
2987
)
122
Oscherwitz
 
J
Gribbin
 
TE
Cease
 
KB
A CD20 tandem-epitope immunogen elicits antibody in mice that binds murine cell surface CD20 and depletes splenic B cells in vivo.
Mol Immunol
2010
, vol. 
47
 
7
(pg. 
1484
-
1491
)
123
Lo Iacono
 
M
Cavallo
 
F
Quaglino
 
E
, et al. 
A limited autoimmunity to p185neu elicited by DNA and allogeneic cell vaccine hampers the progression of preneoplastic lesions in HER-2/NEU transgenic mice.
Int J Immunopathol Pharmacol
2005
, vol. 
18
 
2
(pg. 
351
-
363
)
124
Pupa
 
SM
Iezzi
 
M
Di Carlo
 
E
, et al. 
Inhibition of mammary carcinoma development in HER-2/neu transgenic mice through induction of autoimmunity by xenogeneic DNA vaccination.
Cancer Res
2005
, vol. 
65
 
3
(pg. 
1071
-
1078
)
125
Roskrow
 
MA
Dilloo
 
D
Suzuki
 
N
Zhong
 
W
Rooney
 
CM
Brenner
 
MK
Autoimmune disease induced by dendritic cell immunization against leukemia.
Leuk Res
1999
, vol. 
23
 
6
(pg. 
549
-
557
)
126
Ludewig
 
B
Ochsenbein
 
AF
Odermatt
 
B
Paulin
 
D
Hengartner
 
H
Zinkernagel
 
RM
Immunotherapy with dendritic cells directed against tumor antigens shared with normal host cells results in severe autoimmune disease.
J Exp Med
2000
, vol. 
191
 
5
(pg. 
795
-
804
)

Author notes

*

S.M.A. and C.P.M.D. contributed equally to this study as first authors.

P.K.D. and M.H.K. contributed equally to this study as senior authors.

Sign in via your Institution